Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
FEBS J ; 288(9): 2989-3009, 2021 05.
Article in English | MEDLINE | ID: mdl-33128817

ABSTRACT

ATP synthase, a highly conserved protein complex that has a subunit composition of α3 ß3 γδεab2 c8-15 for the bacterial enzyme, is a key player in supplying energy to living organisms. This protein complex consists of a peripheral F1 sector (α3 ß3 γδε) and a membrane-integrated Fo sector (ab2 c8-15 ). Structural analyses of the isolated protein components revealed that, remarkably, the C-terminal domain of its ε-subunit seems to adopt two dramatically different structures, but the physiological relevance of this conformational change remains largely unknown. In an attempt to decipher this, we developed a high-throughput in vivo protein photo-cross-linking analysis pipeline based on the introduction of the unnatural amino acid into the target protein via the scarless genome-targeted site-directed mutagenesis technique, and probing the cross-linked products via the high-throughput polyacrylamide gel electrophoresis technique. Employing this pipeline, we examined the interactions involving the C-terminal helix of the ε-subunit in cells living under a variety of experimental conditions. These studies enabled us to uncover that the bacterial ATP synthase exists as an equilibrium between the 'inserted' and 'noninserted' state in cells, maintaining a moderate but significant level of net ATP synthesis when shifting to the former upon exposing to unfavorable energetically stressful conditions. Such a mechanism allows the bacterial ATP synthases to proportionally and instantly switch between two reversible functional states in responding to changing environmental conditions. Importantly, this high-throughput approach could allow us to decipher the physiological relevance of protein-protein interactions identified under in vitro conditions or to unveil novel physiological context-dependent protein-protein interactions that are unknown before.


Subject(s)
Protein Conformation , Protein Subunits/genetics , Proteins/ultrastructure , Proton-Translocating ATPases/ultrastructure , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Amino Acid Sequence/genetics , Amino Acids/genetics , Energy Metabolism/genetics , Escherichia coli/enzymology , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Mutagenesis, Site-Directed , Proteins/genetics , Proton-Translocating ATPases/genetics , ATPase Inhibitory Protein
2.
Biochem Biophys Res Commun ; 522(2): 374-380, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31761325

ABSTRACT

The F-ATP synthase is an essential enzyme in mycobacteria, including the pathogenic Mycobacterium tuberculosis. Several new compounds in the TB-drug pipeline target the F-ATP synthase. In light of the importance and pharmacological attractiveness of this novel antibiotic target, tools have to be developed to generate a recombinant mycobacterial F1FO ATP synthase to achieve atomic insight and mutants for mechanistic and regulatory understanding as well as structure-based drug design. Here, we report the first genetically engineered, purified and enzymatically active recombinant M. smegmatis F1FO ATP synthase. The projected 2D- and 3D structures of the recombinant enzyme derived from negatively stained electron micrographs are presented. Furthermore, the first 2D projections from cryo-electron images are revealed, paving the way for an atomic resolution structure determination.


Subject(s)
Proton-Translocating ATPases/metabolism , Recombinant Proteins/metabolism , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Hydrolysis , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Proton-Translocating ATPases/isolation & purification , Proton-Translocating ATPases/ultrastructure , Recombinant Proteins/isolation & purification
3.
Annu Rev Biochem ; 88: 515-549, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30901262

ABSTRACT

F1Fo ATP synthases produce most of the ATP in the cell. F-type ATP synthases have been investigated for more than 50 years, but a full understanding of their molecular mechanisms has become possible only with the recent structures of complete, functionally competent complexes determined by electron cryo-microscopy (cryo-EM). High-resolution cryo-EM structures offer a wealth of unexpected new insights. The catalytic F1 head rotates with the central γ-subunit for the first part of each ATP-generating power stroke. Joint rotation is enabled by subunit δ/OSCP acting as a flexible hinge between F1 and the peripheral stalk. Subunit a conducts protons to and from the c-ring rotor through two conserved aqueous channels. The channels are separated by ∼6 Šin the hydrophobic core of Fo, resulting in a strong local field that generates torque to drive rotary catalysis in F1. The structure of the chloroplast F1Fo complex explains how ATPase activity is turned off at night by a redox switch. Structures of mitochondrial ATP synthase dimers indicate how they shape the inner membrane cristae. The new cryo-EM structures complete our picture of the ATP synthases and reveal the unique mechanism by which they transform an electrochemical membrane potential into biologically useful chemical energy.


Subject(s)
Adenosine Triphosphate/metabolism , Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Bacteria/enzymology , Bacteria/metabolism , Chloroplast Proton-Translocating ATPases/chemistry , Chloroplast Proton-Translocating ATPases/metabolism , Chloroplast Proton-Translocating ATPases/ultrastructure , Chloroplasts/enzymology , Cryoelectron Microscopy , Eukaryota/enzymology , Eukaryota/metabolism , Humans , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/ultrastructure , Protein Conformation , Protein Subunits , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure
4.
Elife ; 82019 02 06.
Article in English | MEDLINE | ID: mdl-30724163

ABSTRACT

ATP synthases produce ATP from ADP and inorganic phosphate with energy from a transmembrane proton motive force. Bacterial ATP synthases have been studied extensively because they are the simplest form of the enzyme and because of the relative ease of genetic manipulation of these complexes. We expressed the Bacillus PS3 ATP synthase in Eschericia coli, purified it, and imaged it by cryo-EM, allowing us to build atomic models of the complex in three rotational states. The position of subunit ε shows how it is able to inhibit ATP hydrolysis while allowing ATP synthesis. The architecture of the membrane region shows how the simple bacterial ATP synthase is able to perform the same core functions as the equivalent, but more complicated, mitochondrial complex. The structures reveal the path of transmembrane proton translocation and provide a model for understanding decades of biochemical analysis interrogating the roles of specific residues in the enzyme.


Subject(s)
Bacillus/enzymology , Protein Conformation , Protein Subunits/chemistry , Proton-Translocating ATPases/ultrastructure , Adenosine Triphosphate/chemistry , Cryoelectron Microscopy , Models, Molecular , Molecular Conformation , Proton-Translocating ATPases/chemistry
5.
Nat Commun ; 10(1): 626, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733444

ABSTRACT

Many Gram-negative bacteria, including causative agents of dysentery, plague, and typhoid fever, rely on a type III secretion system - a multi-membrane spanning syringe-like apparatus - for their pathogenicity. The cytosolic ATPase complex of this injectisome is proposed to play an important role in energizing secretion events and substrate recognition. We present the 3.3 Å resolution cryo-EM structure of the enteropathogenic Escherichia coli ATPase EscN in complex with its central stalk EscO. The structure shows an asymmetric pore with different functional states captured in its six catalytic sites, details directly supporting a rotary catalytic mechanism analogous to that of the heterohexameric F1/V1-ATPases despite its homohexameric nature. Situated at the C-terminal opening of the EscN pore is one molecule of EscO, with primary interaction mediated through an electrostatic interface. The EscN-EscO structure provides significant atomic insights into how the ATPase contributes to type III secretion, including torque generation and binding of chaperone/substrate complexes.


Subject(s)
Cryoelectron Microscopy/methods , Proton-Translocating ATPases/metabolism , Proton-Translocating ATPases/ultrastructure , Type III Secretion Systems/metabolism , Type III Secretion Systems/ultrastructure , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , Protein Structure, Secondary
6.
Methods Mol Biol ; 1805: 51-71, 2018.
Article in English | MEDLINE | ID: mdl-29971712

ABSTRACT

F-type adenosine triphosphate (ATP) synthase is a membrane-bound macromolecular complex, which is responsible for the synthesis of ATP, the universal energy source in living cells. This enzyme uses the proton- or sodium-motive force to power ATP synthesis by a unique rotary mechanism and can also operate in reverse, ATP hydrolysis, to generate ion gradients across membranes. The F1Fo-ATP synthases from bacteria consist of eight different structural subunits, forming a complex of ∼550 kDa in size. In the bacterium Ilyobacter tartaricus the ATP synthase has the stoichiometry α3ß3γδεab2c11. This chapter describes a wet-lab working protocol for the purification of several tens of milligrams of pure, heterologously (E. coli-)produced I. tartaricus Na+-driven F1Fo-ATP synthase and its subsequent efficient reconstitution into proteoliposomes. The methods are useful for a broad range of subsequent biochemical and biotechnological applications.


Subject(s)
Biochemistry/methods , Fusobacteria/enzymology , Proton-Translocating ATPases/isolation & purification , Adenosine Triphosphate/metabolism , Chromatography, Affinity , Escherichia coli/enzymology , Hydrolysis , Plasmids/genetics , Proteolipids/metabolism , Proteolipids/ultrastructure , Proton-Translocating ATPases/ultrastructure
7.
Biochem Biophys Res Commun ; 487(2): 477-482, 2017 05 27.
Article in English | MEDLINE | ID: mdl-28431927

ABSTRACT

The F0 c subunit of F0F1 ATPase (F0-c) possesses two membrane-spanning stretches with N- and C-termini exposed to the periplasmic (extracellular) side of the cytoplasmic membrane of E. coli. Although F0-c insertion has been extensively analyzed in vitro by means of protease protection assaying, it is unclear whether such assays allow elucidation of the insertion process faithfully, since the membrane-protected fragment, an index of membrane insertion, is a full-length polypeptide of F0-c, which is the same as the protease-resistant conformation without membrane insertion. We found that the protease-resistant conformation could be discriminated from membrane-insertion by including octyl glucoside on protease digestion. By means of this system, we found that F0-c insertion depends on MPIase, a glycolipozyme involved in membrane insertion, and is stimulated by YidC. In addition, we found that acidic phospholipids PG and CL transform F0-c into a protease-resistant form, while MPIase prevents the acquisition of such a protease-resistant conformation.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/ultrastructure , Lipid Bilayers/chemistry , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/ultrastructure , Mitochondrial Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Cell Membrane/chemistry , Mitochondrial Proton-Translocating ATPases/ultrastructure , Structure-Activity Relationship
8.
Nature ; 521(7551): 237-40, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25707805

ABSTRACT

ATP, the universal energy currency of cells, is produced by F-type ATP synthases, which are ancient, membrane-bound nanomachines. F-type ATP synthases use the energy of a transmembrane electrochemical gradient to generate ATP by rotary catalysis. Protons moving across the membrane drive a rotor ring composed of 8-15 c-subunits. A central stalk transmits the rotation of the c-ring to the catalytic F1 head, where a series of conformational changes results in ATP synthesis. A key unresolved question in this fundamental process is how protons pass through the membrane to drive ATP production. Mitochondrial ATP synthases form V-shaped homodimers in cristae membranes. Here we report the structure of a native and active mitochondrial ATP synthase dimer, determined by single-particle electron cryomicroscopy at 6.2 Å resolution. Our structure shows four long, horizontal membrane-intrinsic α-helices in the a-subunit, arranged in two hairpins at an angle of approximately 70° relative to the c-ring helices. It has been proposed that a strictly conserved membrane-embedded arginine in the a-subunit couples proton translocation to c-ring rotation. A fit of the conserved carboxy-terminal a-subunit sequence places the conserved arginine next to a proton-binding c-subunit glutamate. The map shows a slanting solvent-accessible channel that extends from the mitochondrial matrix to the conserved arginine. Another hydrophilic cavity on the lumenal membrane surface defines a direct route for the protons to an essential histidine-glutamate pair. Our results provide unique new insights into the structure and function of rotary ATP synthases and explain how ATP production is coupled to proton translocation.


Subject(s)
Chlorophyta/enzymology , Protein Subunits/chemistry , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/metabolism , Arginine/metabolism , Cryoelectron Microscopy , Glutamic Acid/metabolism , Histidine/metabolism , Ion Transport , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Models, Molecular , Protein Multimerization , Protein Structure, Secondary , Protein Subunits/metabolism , Proton-Translocating ATPases/metabolism , Protons , Rotation , Water/metabolism
9.
Biochem Biophys Res Commun ; 452(4): 940-4, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25230139

ABSTRACT

ATP synthase (F-ATPase) function depends upon catalytic and rotation cycles of the F1 sector. Previously, we found that F1 ATPase activity is inhibited by the dietary polyphenols, curcumin, quercetin, and piceatannol, but that the inhibitory kinetics of curcumin differs from that of the other two polyphenols (Sekiya et al., 2012, 2014). In the present study, we analyzed Escherichia coli F1 ATPase rotational catalysis to identify differences in the inhibitory mechanism of curcumin versus quercetin and piceatannol. These compounds did not affect the 120° rotation step for ATP binding and ADP release, though they significantly increased the catalytic dwell duration for ATP hydrolysis. Analysis of wild-type F1 and a mutant lacking part of the piceatannol binding site (γΔ277-286) indicates that curcumin binds to F1 differently from piceatannol and quercetin. The unique inhibitory mechanism of curcumin is also suggested from its effect on F1 mutants with defective ß-γ subunit interactions (γMet23 to Lys) or ß conformational changes (ßSer174 to Phe). These results confirm that smooth interaction between each ß subunit and entire γ subunit in F1 is pertinent for rotational catalysis.


Subject(s)
Curcumin/chemistry , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/ultrastructure , Amino Acid Sequence , Binding Sites , Enzyme Activation , Models, Chemical , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
10.
IUBMB Life ; 65(3): 227-37, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23378185

ABSTRACT

Optical microscopy of single F(1) -ATPase and F(o) F(1) -ATP synthases started 15 years ago. Direct demonstration of ATP-driven subunit rotation by videomicroscopy became the new exciting tool to analyze the conformational changes of this enzyme during catalysis. Stimulated by these experiments, technical improvements for higher time resolution, better angular resolution, and reduced viscous drag were developed rapidly. Optics and single-molecule enzymology were entangled to benefit both biochemists and microscopists. Today, several single-molecule microscopy methods are established including controls for the precise nanomanipulation of individual enzymes in vitro. Förster resonance energy transfer, which has been used for simultaneous monitoring of conformational changes of different parts of this rotary motor, is one of them and may become the tool for the analysis of single F(o) F(1) -ATP synthases in membranes of living cells. Here, breakthrough experiments are critically reviewed and challenges are discussed for the future microscopy of single ATP synthesizing enzymes at work.


Subject(s)
Adenosine Triphosphate/biosynthesis , Escherichia coli Proteins/ultrastructure , Escherichia coli/enzymology , Molecular Motor Proteins/ultrastructure , Protein Subunits/chemistry , Proton-Translocating ATPases/ultrastructure , Biocatalysis , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Fluorescence Resonance Energy Transfer , Microscopy, Video , Models, Molecular , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/metabolism , Protein Subunits/metabolism , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Rotation , Thermodynamics
11.
Nature ; 481(7380): 214-8, 2011 Dec 18.
Article in English | MEDLINE | ID: mdl-22178924

ABSTRACT

Ion-translocating rotary ATPases serve either as ATP synthases, using energy from a transmembrane ion motive force to create the cell's supply of ATP, or as transmembrane ion pumps that are powered by ATP hydrolysis. The members of this family of enzymes each contain two rotary motors: one that couples ion translocation to rotation and one that couples rotation to ATP synthesis or hydrolysis. During ATP synthesis, ion translocation through the membrane-bound region of the complex causes rotation of a central rotor that drives conformational changes and ATP synthesis in the catalytic region of the complex. There are no structural models available for the intact membrane region of any ion-translocating rotary ATPase. Here we present a 9.7 Å resolution map of the H(+)-driven ATP synthase from Thermus thermophilus obtained by electron cryomicroscopy of single particles in ice. The 600-kilodalton complex has an overall subunit composition of A(3)B(3)CDE(2)FG(2)IL(12). The membrane-bound motor consists of a ring of L subunits and the carboxy-terminal region of subunit I, which are equivalent to the c and a subunits of most other rotary ATPases, respectively. The map shows that the ring contains 12 L subunits and that the I subunit has eight transmembrane helices. The L(12) ring and I subunit have a surprisingly small contact area in the middle of the membrane, with helices from the I subunit making contacts with two different L subunits. The transmembrane helices of subunit I form bundles that could serve as half-channels across the membrane, with the first half-channel conducting protons from the periplasm to the L(12) ring and the second half-channel conducting protons from the L(12) ring to the cytoplasm. This structure therefore suggests the mechanism by which a transmembrane proton motive force is converted to rotation in rotary ATPases.


Subject(s)
Cryoelectron Microscopy , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Protons , Thermus thermophilus/enzymology , Cell Membrane/metabolism , Ice , Models, Biological , Models, Molecular , Protein Subunits/chemistry , Protein Subunits/metabolism , Proton-Motive Force , Proton-Translocating ATPases/metabolism , Rotation , Structure-Activity Relationship
13.
J Bioenerg Biomembr ; 41(4): 343-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19760172

ABSTRACT

The N-termini of E and H of A1AO ATP synthase have been shown to interact and an NMR structure of N-terminal H1-47 has been solved recently. In order to understand the E-H assembly and the N-terminal structure of E, the truncated construct E1-52 of Methanocaldococcus jannaschii A1AO ATP synthase was produced, purified and the solution structure of E1-52 was determined by NMR spectroscopy. The protein is 60.5 A in length and forms an alpha helix between the residues 8-48. The molecule is amphipathic with a strip of hydrophobic residues, discussed as a possible helix-helix interaction with neighboring subunit H.


Subject(s)
Archaeal Proteins/chemistry , Archaeal Proteins/ultrastructure , Euryarchaeota/enzymology , Magnetic Resonance Spectroscopy/methods , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Amino Acid Sequence , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Protein Subunits
14.
Planta ; 229(5): 1087-98, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19225806

ABSTRACT

The plasma membrane H(+)-ATPase (PM H(+)-ATPase, EC.3.6.1.35) plays a key role in the plant response to environmental stress. In this study, a possible mechanistic link between the PM H(+)-ATPase and salicylic acid (SA)-induced thermotolerance was investigated in pea (Pisum sativum L. cv. NingXia) leaves. The burst of free SA in response to heat acclimation (38 +/- 0.5 degrees C) was observed, and peaks appeared subsequently both in activity and amount of PM H(+)-ATPase in pea leaves during heat acclimation. Similarly, exogenous SA also triggered the two peaks in the room temperature (25 +/- 0.5 degrees C). Paclobutrazol (PAC) was employed to infiltrate onto pea leaves prior to heat acclimation treatment. The results showed that the peaks of both free SA and activity of PM H(+)-ATPase still occurred after the PAC pretreatment. In acquired thermotolerance assessment (malondialdehyde content and degree of wilting), spraying SA and fusicoccin (FC, the activator of PM H(+)-ATPase) separately could protect pea leaves from heat injury. Results from RT-PCR and western blotting analysis indicated that the increase in activity of the PM H(+)-ATPase was due to its transcriptional and translational regulation. The subcellular localizations of PM H(+)-ATPase after the FC or SA pretreatment also showed that the PM H(+)-ATPase is important to maintain the integrity of plasma membrane against the heat stress. Taken together, these results suggest PM H(+)-ATPase is related to the development of SA-induced thermotolerance in pea leaves.


Subject(s)
Adaptation, Physiological/drug effects , Cell Membrane/enzymology , Pisum sativum/enzymology , Plant Leaves/enzymology , Proton-Translocating ATPases/metabolism , Salicylic Acid/pharmacology , Temperature , Acclimatization/drug effects , Cell Membrane/ultrastructure , Cell Membrane Permeability/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Plant/drug effects , Malondialdehyde/metabolism , Pisum sativum/cytology , Pisum sativum/drug effects , Pisum sativum/ultrastructure , Plant Leaves/cytology , Plant Leaves/drug effects , Plant Leaves/ultrastructure , Protein Transport/drug effects , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Triazoles/pharmacology
15.
Prog Biophys Mol Biol ; 99(1): 20-41, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19049812

ABSTRACT

Peter D. Mitchell, who was awarded the Nobel Prize in Chemistry 30 years ago, in 1978, formulated the chemiosmotic theory of oxidative phosphorylation. This review initially analyzes the major aspects of this theory, its unresolved problems, and its modifications. A new physico-chemical mechanism of energy transformation and coupling of oxidation and phosphorylation is then suggested based on recent concepts regarding proteins, including ATPases that work as molecular motors, and acidic lipids that act as hydrogen ion (H(+)) carriers. According to this proposed mechanism, the chemical energy of a redox substrate is transformed into nonequilibrium states of electron-transporting chain (ETC) coupling proteins. This leads to nonequilibrium pumping of H(+) into the membrane. An acidic lipid, cardiolipin, binds with this H(+) and carries it to the ATP-synthase along the membrane surface. This transport generates gradients of surface tension or electric field along the membrane surface. Hydrodynamic effects on a nanolevel lead to rotation of ATP-synthase and finally to the release of ATP into aqueous solution. This model also explains the generation of a transmembrane protonmotive force that is used for regulation of transmembrane transport, but is not necessary for the coupling of electron transport and ATP synthesis.


Subject(s)
Lipids/chemistry , Models, Chemical , Models, Molecular , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Acids , Enzyme Activation , Nobel Prize , Oxidation-Reduction , Phosphorylation
16.
J Bioenerg Biomembr ; 40(4): 269-79, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18846414

ABSTRACT

Sulfate is a partial inhibitor at low and a non-essential activator at high [ATP] of the ATPase activity of F(1). Therefore, a catalytically-competent ternary F(1) x ATP x sulfate complex can be formed. In addition, the ANS fluorescence enhancement driven by ATP hydrolysis in submitochondrial particles is also stimulated by sulfate, clearly showing that the ATP hydrolysis in its presence is coupled to H(+) translocation. However, sulfate is a strong linear inhibitor of the mitochondrial ATP synthesis. The inhibition was competitive (K (i) = 0.46 mM) with respect to Pi and mixed (K (i) = 0.60 and K'(i) = 5.6 mM) towards ADP. Since it is likely that sulfate exerts its effects by binding at the Pi binding subdomain of the catalytic site, we suggest that the catalytic site involved in the H(+) translocation driven by ATP hydrolysis has a more open conformation than the half-closed one (beta(HC)), which is an intermediate in ATP synthesis. Accordingly, ATP hydrolysis is not necessarily the exact reversal of ATP synthesis.


Subject(s)
Adenosine Triphosphate/chemistry , Adenosine Triphosphate/chemical synthesis , Mitochondria, Heart/enzymology , Models, Chemical , Models, Molecular , Myocardium/enzymology , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Sulfates/chemistry , Animals , Binding Sites , Cattle , Computer Simulation , Enzyme Activation , Enzyme Stability , Hydrolysis , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protons
17.
Biochem Biophys Res Commun ; 377(1): 191-4, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18835248

ABSTRACT

We have developed a novel micro-mixer using a biological molecular ATP motor. The micro-mixer was constructed from arrays of chromatophore-embedded delta-free F(0)F(1)-ATPases, where the delta-free F(1) part acted as a rotator to mix solutions, and the F(0) part was driven by light. Confocal microscope studies indicated that the micro-mixer did not touch directly on the fibrin labeled with FITC. The nanomechanical force generated by the motor induced drug movement in the solution and accelerated the fibrinolysis process. All results strongly suggest that the micro-mixers generated a nanomechanical force which accelerated the fibrinolysis process in the presence of lower concentrations of lumbrokinase.


Subject(s)
Fibrin/chemistry , Fibrinolysis , Nanotechnology , Proton-Translocating ATPases/chemistry , Fibrin/radiation effects , Fibrin/ultrastructure , Fibrinolysis/radiation effects , Fluorescein-5-isothiocyanate/chemistry , Glass/chemistry , Light , Microscopy, Confocal , Proton-Translocating ATPases/radiation effects , Proton-Translocating ATPases/ultrastructure , Surface Properties , Thrombolytic Therapy
18.
J Bioenerg Biomembr ; 40(4): 257-67, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18958608

ABSTRACT

The role of the integral inner membrane subunit e in self-association of F(0)F(1)ATP synthase from bovine heart mitochondria was analyzed by in situ limited proteolysis, blue native PAGE/iterative SDS-PAGE, and LC-MS/MS. Selective degradation of subunit e, without disrupting membrane integrity or ATPase capacity, altered the oligomeric distribution of F(0)F(1)ATP synthase, by eliminating oligomers and reducing dimers in favor of monomers. The stoichiometry of subunit e was determined by a quantitative MS-based proteomics approach, using synthetic isotope-labelled reference peptides IAQL*EEVK, VYGVGSL*ALYEK, and ELAEAQEDTIL*K to quantify the b, gamma and e subunits, respectively. Accuracy of the method was demonstrated by confirming the 1:1 stoichiometry of subunits gamma and b. Altogether, the results indicate that the integrity of a unique copy of subunit e is essential for self-association of mammalian F(0)F(1)ATP synthase.


Subject(s)
Adenosine Triphosphate/chemistry , Mitochondria, Heart/enzymology , Models, Chemical , Models, Molecular , Myocardium/enzymology , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Animals , Binding Sites , Cattle , Computer Simulation , Enzyme Activation , Enzyme Stability , Protein Binding , Protein Conformation , Protein Subunits/chemistry
19.
J Bioenerg Biomembr ; 40(4): 359-69, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18770013

ABSTRACT

We studied for the first time the ATP-synthase complex from shrimp as a model to understand the basis of crustacean bioenergetics since they are exposed to endogenous processes as molting that demand high amount of energy. We analyzed the cDNA sequence of two subunits of the Fo sector from mitochondrial ATP-synthase in the white shrimp Litopenaeus vannamei. The nucleus encoded atp9 subunit presents a 773 bp sequence, containing a signal peptide sequence only observed in crustaceans, and the mitochondrial encoded atp6 subunit presents a sequence of 675 bp, and exhibits high identity with homologous sequences from invertebrate species. ATP9 and ATP6 protein structural models interaction suggest specific functional characteristics from both proteins in the mitochondrial enzyme. Differences in the steady-state mRNA levels of atp9 and atp6 from five different tissues correlate with tissue function. Moreover, significant changes in the mRNA levels of both subunits at different molt stages were detected. We discussed some insights about the enzyme structure and the regulation mechanisms from both ATP-synthase subunits related to the energy requirements of shrimp.


Subject(s)
Cell Nucleus/enzymology , Mitochondria/enzymology , Models, Chemical , Models, Molecular , Penaeidae/enzymology , Proton-Translocating ATPases , Amino Acid Sequence , Animals , Base Sequence , Computer Simulation , Gene Expression Profiling , Molecular Sequence Data , Protein Conformation , Protein Subunits , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/ultrastructure , RNA, Messenger/genetics
20.
Biophys J ; 95(10): 4979-87, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18723591

ABSTRACT

The F(O)F(1)-ATPase is a rotary molecular motor. Driven by ATP-hydrolysis, its central shaft rotates in 80 degrees and 40 degrees steps, interrupted by catalytic and ATP-waiting dwells. We recorded rotations and halts by means of microvideography in laboratory coordinates. A correlation with molecular coordinates was established by using an engineered pair of cysteines that, under oxidizing conditions, formed zero-length cross-links between the rotor and the stator in an orientation as found in crystals. The fixed orientation coincided with that of the catalytic dwell, whereas the ATP waiting dwell was displaced from it by +40 degrees . In crystals, the convex side of the cranked central shaft faces an empty nucleotide binding site, as if holding it open for arriving ATP. Functional studies suggest that three sites are occupied during a catalytic dwell. Our data imply that the convex side faces a nucleotide-occupied rather than an empty site. The enzyme conformation in crystals seems to differ from the conformation during either dwell of the active enzyme. A revision of current schemes of the mechanism is proposed.


Subject(s)
Adenosine Triphosphate/chemistry , Models, Chemical , Models, Molecular , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/ultrastructure , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Computer Simulation , Crystallography , Protein Conformation , Rotation , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...